13

Click here to load reader

NOX enzymes: potential target for the treatment of acute lung injury

Embed Size (px)

Citation preview

Page 1: NOX enzymes: potential target for the treatment of acute lung injury

MULTI-AUTHOR REVIEW

NOX enzymes: potential target for the treatment of acute lunginjury

Stephanie Carnesecchi • Jean-Claude Pache •

Constance Barazzone-Argiroffo

Received: 18 April 2012 / Revised: 18 April 2012 / Accepted: 20 April 2012 / Published online: 13 May 2012

� Springer Basel AG 2012

Abstract Acute lung injury (ALI) and its more severe

form, acute respiratory distress syndrome (ARDS), is char-

acterized by acute inflammation, disruption of the alveolar-

capillary barrier, and in the organizing stage by alveolar

pneumocytes hyperplasia and extensive lung fibrosis. The

cellular and molecular mechanisms leading to the develop-

ment of ALI/ARDS are not completely understood, but there

is evidence that reactive oxygen species (ROS) generated by

inflammatory cells as well as epithelial and endothelial cells

are responsible for inflammatory response, lung damage, and

abnormal repair. Among all ROS-producing enzymes, the

members of NADPH oxidases (NOXs), which are widely

expressed in different lung cell types, have been shown to

participate in cellular processes involved in the maintenance

of lung integrity. It is not surprising that change in NOXs’

expression and function is involved in the development of

ALI/ARDS. In this context, the use of NOX inhibitors could

be a possible therapeutic perspective in the management of

this syndrome. In this article, we summarize the current

knowledge concerning some cellular aspects of NOXs

localization and function in the lungs, consider their contri-

bution in the development of ALI/ARDS and discuss the

place of NOX inhibitors as potential therapeutical target.

Keywords NOX enzymes � Acute lung injury �Acute respiratory distress syndrome

Abbreviations

ALI Acute lung injury

ARDS Acute respiratory distress syndrome

BAL Bronchoalveolar lavage

ROS Reactive oxygen species

HM Hyaline membranes

LPS Lipopolysaccharide

TNF-a Tumor necrosis factor-aICAM-1 Intracellular adhesion molecule-1

EC Endothelial cells

TGF-b1 Transforming growth factor-b1

IPF Idiopathic pulmonary fibrosis

IRF-3 Interferon regulatory factor-3

AP1 Activator protein 1

NF-jB Nucleor factor jB

MCP-1 Monocyte chemotactic protein-1

MV Mechanical ventilation

N.D. Not determined

Introduction

Acute lung injury (ALI) and acute respiratory distress

syndrome (ARDS), which is the most severe form, is

associated with a high mortality (50–80 %). ALI/ARDS

affects a large number of patients entering intensive care

units and is defined by bilateral pulmonary infiltrates on

chest radiograph, hypoxemic respiratory failure measured

by a partial pressure of arterial oxygen (PaO2)/fraction of

inspired oxygen (FiO2) ratio (PaO2/FiO2 \ 300 mmHg for

ALI and \200 mmHg for ARDS) with normal hydrostatic

pressure corresponding to the absence of left heart failure.

Acute respiratory distress syndrome can occur with several

diseases either associated with those causing direct lung

injury such as pneumonia, gastric aspiration or toxic

S. Carnesecchi (&) � C. Barazzone-Argiroffo

Department of Pediatrics/Pathology and Immunology,

Centre Medical Universitaire, 1 rue Michel Servet,

1211 Geneva, Switzerland

e-mail: [email protected]

J.-C. Pache

Department of Pathology and Immunology, Centre Medical

Universitaire, 1 rue Michel Servet, 1211 Geneva, Switzerland

Cell. Mol. Life Sci. (2012) 69:2373–2385

DOI 10.1007/s00018-012-1013-6 Cellular and Molecular Life Sciences

123

Page 2: NOX enzymes: potential target for the treatment of acute lung injury

inhalation, or indirect injury such as sepsis or severe burn.

The heterogeneity of causes and the complexity of clinical

histopathological and radiographic manifestations make

the study of ARDS pathogenesis and the test of new

therapeutics difficult. Indeed, ARDS follows most often a

progressive course characterized by two distinct stages.

The acute (or exudative) stage includes the disruption of

the alveolar-capillary barrier, pulmonary edema, accumu-

lation of protein-rich fluid into the interstitium, alveolar

space, and diffuse inflammation. The later organizing stage

occurs with the resolution of pulmonary edema and lung

inflammation. During this phase, alveolar pneumocytes

hyperplasia and fibroblast proliferation lead to disordered

collagen deposition and extensive lung fibrosis.

Although the elucidation of the cellular and molecular

mechanisms involved in the pathogenesis of ALI/ARDS

remain unclear and complex, there is evidence that reactive

oxygen species (ROS) contribute to the initiation of

endothelial damage characteristic of ARDS and are

responsible for most of clinical symptoms of this syndrome

[1]. Indeed, a large amount of ROS, which are found in

broncho alveolar lavages (BAL) of ARDS patients are

produced mainly by alveolar macrophages, neutrophils,

lung endothelial, and epithelial cells. These ROS can alter

gene and protein function. Among several ROS-producing

enzymes, NADPH oxidase (NOX) enzymes, which are

membrane-bound complexes catalyzing the reduction of

molecular oxygen (O2) to superoxide (O2-) [2], are

involved in principal clinical manifestations of ALI/ARDS

[1, 3]. The first NOX has been described in phagocytes and

is a complex that includes a catalytic subunit gp91phox

called NOX2 associated with p22phox and cytosolic regu-

latory subunits such p47phox, p67phox and small GTPase

(RAC1 or RAC 2), required for NOX activation and gen-

eration of superoxide [2, 4]. Recently, structural

homologues of the phagocyte NOX enzyme were identi-

fied, such as NOX1-3-4-5, DUOX1, and DUOX2. Despite

their similar structure and enzymatic function, NOX

enzymes differ in their mechanisms of activation, which

depend on the recruitment of membrane or/and cytosolic

regulatory subunits such as p22phox, p47phox, p67phox,

NOXO1, NOXA1, and RAC [2]. The NOX isoforms,

which are expressed in a variety of lung cell types [5],

participate in several cellular processes [6] and are

involved in lung pathological situations such as ALI/

ARDS, cancer, fibrosis, pulmonary hypertension, and

obstructive lung disorders such as emphysema, asthma, and

cystic fibrosis [5, 7–10].

In the present article, we will focus on the contribution of

NOX enzymes in the development of ALI/ARDS. We will

first briefly describe some cellular aspects of NOX locali-

zation and function in the lungs. In the second part, we will

review the current knowledge concerning the role of NOX-

dependent ROS production in the pathogenesis of ALI/

ARDS and particularly its involvement in some clinical

aspects of this disease. In the third part, we will discuss their

therapeutic potential in the management of ARDS/ALI.

Cellular expression and function of NOX enzymes

in lungs

The lung, of which the principal function is to deliver

oxygen to tissues, is widely exposed to deleterious envi-

ronmental factors including virus, bacteria, irritants and

allergens, and possesses a potent innate defense system.

This defense system not only uses the phagocyte NOX

system to eliminate these dangers through oxidative killing

[6, 11] but also in regulating cell-signaling pathways

involved in host defense mechanisms, cell proliferation,

migration, and/or differentiation [4, 12]. Several studies

have shown that NOX enzymes are expressed in lungs,

both in mice and in humans. The amount of the different

NOX isoforms depends on the cell types and also on the

species. A high amount of NOX2 [13, 14] and DUOX1/2

mRNA [15, 16] as well as NOX1 [17, 18] and NOX4 [7, 8,

13, 18] are detected in lungs. In addition to the expression

of NOX2 in alveolar macrophages and other inflammatory

cell types, NOX isoforms have been detected in different

lung cell types such as alveolar epithelial and endothelial

cells, fibroblasts, smooth muscle cells, and airway epithe-

lial cells [19, 20]. The cell-type-dependent NOX

expression in lungs suggests their specific participation in

some aspect of physiological and pathological functions

including host defenses, proliferation, migration, and/or

differentiation. The specific lung expression and function

of NOX enzymes are summarized in the Table 1.

Thus, according to the physiological contribution of

NOX enzymes in tissue repair and/or remodeling, we could

envisage that the modulation of their expression and acti-

vation in different lung cell types contributes to the

development of lung diseases such as ALI/ARDS. We will

first describe the pathogenesis of ALI/ARDS and then the

role of ROS-dependent NOX enzymes in different animal

models of ARDS/ALI.

Histopathology and pathogenesis of ALI/ARDS

In spite of the scarce knowledge concerning the mecha-

nisms involved in the pathogenesis of ALI/ARDS,

histological analysis of lung sections from ARDS at dif-

ferent stages suggests that lung modifications occurring

during this disease follow a scheduled time course and can

be divided into three time-dependent phases: acute (or

exudative), proliferative, and fibrosis [61].

2374 S. Carnesecchi et al.

123

Page 3: NOX enzymes: potential target for the treatment of acute lung injury

The acute phase begins after the initial injury; most cells

composing the alveolar septa undergo either apoptosis or

necrosis, and inflammatory cells mainly represented by the

neutrophils, invade alveolar walls and lumens. The number

of neutrophils increases fast as they travel via the inter-

stitium into the airspaces. This is facilitated by the

disruption of the vascular structures, which occurs when

neutrophils and fibrin plugs occlude the capillaries

(Fig. 1a, b). Hyaline membranes (HM) made of fibrin,

proteins and cellular debris; accumulate along the alveolar

walls (Fig. 1a, b). By electron microscopy, all stages of

epithelial degeneration can be observed from slight cyto-

plasmic swelling to huge blister formation and total

destruction of the epithelial lining [62]. In parallel, the

endothelial cell layer is often irregular because of cyto-

plasmic swelling and large vacuoles. Endothelial defects

Table 1 Summary of NOX enzymes localization, activation, and function in lung cells

NOX

isoforms

Expression Stimuli Function Species References

NOX1 Endothelial cells FGF-b, VEGF Vascular cell growth M, H [21]

FGF-b, VEGF Angiogenesis M, H [21]

Hyperoxia Cell death M [17]

Alveolar epithelial

cells

TNF-a, hyperoxia Cell death M [17, 22]

Hypoxia HIF-a signaling H [23, 24]

Growth factors, HIPK2 depletion Proliferation M, H [25, 26]

Fibroblasts N.D N.D M Personal

data

Vascular smooth

muscle cells

– N.D R [27]

NOX2 Endothelial cells Hyperoxia Cell migration H [28, 29]

Ischemia and High K?, hypoxia Oxygen sensing B, M [30–33]

LPS, TNF-a TLR2 crosstalk M [34]

Neuro-epithelial

cells

Hypoxia Chemoreceptor O2 sensing H, R,

Ra

[35]

Macrophages/

neutrophils

TNF-a, LPS influenza A virus Anti-microbial host defense/innate

immune response

M, H [34, 36–41]

Chronic fine particulate TLR4 crosstalk, NF-jB activation M [42]

NOX3 Endothelial cells – TLR4 crosstalk M [10]

Hyperoxia Cell integrity M [43]

NOX4 Endothelial cells Hyperoxia Cell migration H [28, 44]

Alveolar epithelial

cells

Bleomycin, TGF-b1, fine particles Cell death M, H [7, 45]

Smooth muscle

cells

TGF-b1 Proliferation M, H [18, 46, 47]

Differentiation R [27]

Fibroblasts/

myofibroblasts

Bleomycin, TGF-b1, radiation Differentiation/activation M, H [7, 8, 48, 49]

Hypoxia Proliferation H [50]

DUOX1 Bronchial cells Pseudomonas aeruginosa, LPO, IL-4, IL-13

cytokines, and cigarette smoke

Host defense H [15, 19, 20,

51–54]

PMA, human neutrophil elastase Mucin expression H [55]

ATP Cellular migration H [56]

– H? production and secretion H [57]

LPS Cell proliferation M [58]

Alveolar epithelial

cells

Hormone mixture Differentiation H [59]

DUOX2 Bronchial cells IFN-c Host defense H [19, 60]

– H? production and secretion H [57]

Alveolar epithelial

cells

– N.D M Personal

data

M mouse, H human, R rat, Ra rabbit, B bovine, PMA phorbol 12-myristate 13-acetate, HIPK2 homeo domain-interacting protein kinase-2, LPOlactoperoxidase, ATP adenosine triphosphate, ANPH atrial natriuretic peptide hormone, N.D not determined

NOX enzymes and acute lung injury 2375

123

Page 4: NOX enzymes: potential target for the treatment of acute lung injury

are covered by fibrin or microthrombi, which completely

obliterate the capillaries (Fig. 1b, enlarged insert). Con-

cerning the late exudative stage, the epithelial lining is thin

and covered by laminated paucicellular HM. The alveolar

septa are enlarged and contain numerous inflammatory

cells.

After approximately 7 days, the organizing or prolifera-

tive stage is observed with increased interstitial cellularity.

The number of large cuboidal cells, which resemble epi-

thelial type II cells and might represent a stem cell population

of the lung (Fig. 1c and d), increases strikingly. The com-

position of the interstitial cells changes; neutrophils are

partly replaced by macrophages, lymphocytes, and plasma

cells. The interstitium is organized by the proliferation of

connective tissue, persisting edema, and convolutes of loose

fibrous tissue without capillaries. There is a strong diminu-

tion of the microvasculature that is sometimes compressed

by the surrounding tissue. Myofibroblasts that express

vimentin and a-smooth muscle actin (a-SMA) are progres-

sively observed in interstitium and then in airspaces, with a

maximum in the early proliferative stage [63]. The late

proliferative phase shows easily identifiable proliferating

intra-septal and/or intra-alveolar myofibroblasts (airspace

fibroplasia) and production of new matrix substances with

the doubling of lung collagen in 2 weeks.

After a few more days, the fibrotic phase shows wide

connective tissue area interspread between alveolar septa.

Bulk tissue masses formed by folded up septa and col-

lapsed alveoli surround unusually wide airspaces, which

originated mostly from widened alveolar ducts or respira-

tory bronchioles. Some airspace is enlarged due to tissue

destruction. The histology is characterized by enlarged

fibrotic septa and laminated intra-alveolar fibrosis.

ROS are increasingly considered key substances in the

initiation of endothelial damage characteristic of ARDS

and are responsible for most of the clinical symptoms of

this syndrome. There are several causes that increase oxi-

dative stress in ARDS such as breathing high inspiratory

oxygen concentration. However, the large majority of

oxidants are generated by phagocytic cells transmigrating

into the lungs. Neutrophils are crucial since they appear

early in histological specimens and are strongly increased

in the BAL. They release many inflammatory mediators

that include chemokines, cytokines, and proteases [61].

EII

HM

E

HMTN

En

A B

C D

MF

EII

IN

EII

TN

En

Fig. 1 Histological hallmarks of acute respiratory distress syndrome

during the exudative and the proliferative phases. a, b Lung sections

stained with hematoxylin and eosin (H&E) obtained from biopsy of

ARDS subjects during the exudative phase. a Deposition of hyaline

membranes (HM) on the epithelial side of the basement membrane.

At this stage, the presence of detached epithelial type II cells from the

alveolar wall (EII) is also apparent. b The presence of the interstitial

edema (E). The necrosis of endothelial cells (En) and the formation of

thrombus associated with the margination of neutrophil (TN) are also

obvious at this stage. Original magnification 9400 (a and b), 9500

for enlarged insert. c, d Lung sections stained with (H&E) obtained

from biopsy of ARDS subjects during the proliferative phase. c, d The

evident hyperplasia of epithelial type II cells (EII) and an extended

zone of interstitial (IN) fibro-proliferation. Note the presence of

myofibroblasts in the parenchyma (MF). Original magnifications

9200 (c), 9400 (d)

2376 S. Carnesecchi et al.

123

Page 5: NOX enzymes: potential target for the treatment of acute lung injury

The activation of the neutrophils may occur at remote sites

and/or by circulating cytokines, resulting in ROS release

and increased pulmonary vascular permeability. However,

neutrophils are not mandatory for the development of

ARDS, because it can occur in neutropenic patients. Other

key initiators of the pulmonary inflammation in ARDS are

the circulating inflammatory mediators (e.g., TNF-a, IL-

1b, IL-6, IL-8, leukotrienes), as well as changes in the

coagulation system. Alveolar edema resulting from endo-

thelial dysfunction and loss of epithelial integrity reduces

the barrier function. The edema is even increased by the

loss of type II pneumocytes that normally promote fluid

transport out of the alveolus through their apical sodium

pumps. Early loss of surfactant is explained by the damage

of type II epithelial cells, which produce surfactant and by

its neutralization by protein-rich edema fluid. This con-

tributes to alveolar collapse, intrapulmonary shunt, and

hypoxemia. The hypoxic pulmonary vasoconstriction is

impaired by endothelial and smooth muscle cell dysfunc-

tion. This may, with the association of microthrombi,

contribute to the development of secondary pulmonary

hypertension.

Role of NOX enzymes in ALI/ARDS

Whereas the mechanisms that initiate ALI/ARDS remain

unclear, there is some evidence that ROS generated by

NOX enzymes participate in the pathogenesis of this syn-

drome. The study of NOX enzyme contribution in the

patho-mechanisms of ALI/ARDS in human is difficult due

to the heterogeneity of causes and the paucity of biological

samples (biopsies, autopsies, BALF). Animal models

reproduce major clinical features of ALI/ARDS observed

in humans including the loss of the alveolar-capillary

barrier with the damage of both epithelial and endothelial

cells and the inflammatory cell influx. In this way, all these

models provide key elements to study the role of the NOX

family during ALI/ARDS. In the next part, we will

examine their involvement in different mouse models of

ALI/ARDS that mimic neutrophilic infiltration and lung

injury in sepsis-like models and the damage of the alveolar-

capillary barrier.

Lung inflammation and injury

Histological analysis of lung sections from ARDS patients

as well as BAL obtained in the acute phase of the disease

show massive accumulation of neutrophils [64] and most

acute lung injuries induced in animal models are neutro-

phil-dependent [65–67]. These inflammatory cells produce

high levels of ROS, which are thought to increase the

inflammatory processes and tissue injury in septic shock

syndrome [68–70]. In addition, ROS participates in the

modulation of cell-signaling pathways that activate tran-

scription factor of redox-sensitive pro-inflammatory

mediators such as NF-jB [71, 72]. The studies concerning

the effect of ROS generated by NOX enzymes in acute

inflammatory responses and lung injury following Esche-

richia coli and lipopolysaccharide (LPS) challenges in

mice are controversial [37, 71, 73, 74]. Indeed, some

studies have shown that the absence of p47phox (a regula-

tory subunit essential for NOX2 activation) did not

contribute to LPS-induced lung damage, vascular leakage,

and infiltration of neutrophils and monocytes in mice [75].

Swain et al. [76] did not observe any improvement of

pulmonary lung injury in gp91phox-deficient mice during

pneumocystis pneumonia. By contrast, it has been dem-

onstrated that LPS-induced inflammation and lung injury

was inhibited but also in some cases increased in NOX-

deficient mouse models. The absence of p47phox and

gp91phox has been associated with enhanced inflammatory

gene expression, lung neutrophil recruitment, and mouse

survival after LPS challenge [77, 78]. On the other hand,

LPS-induced lung inflammation was reduced in mice

deficient for Nrf2, a regulator of antioxidant defenses, in

absence of p47phox or gp91phox [37]. Moreover, ROS pro-

duction restricted to macrophages from Nrf2-deficient mice

was blunted by the absence of gp91phox after LPS challenge

[37]. Similarly, inflammatory response induced by live

Escherichia coli or LPS was reduced in lung tissues of

p47phox- and gp91phox-deficient mice [71, 73]. Sadikot et al.

[74] has reported that NF-jB activation and TNF-a levels

were decreased in p47phox-deficient mice after Pseudomo-

nas aeruginosa infection and a recent study showed that

ROS generated by NOX2 in neutrophils were involved in

TNF-a-induced acute lung injury [38] and participated in

inflammatory response through the activation of NF-jB

[36]. These results support the notion that ROS generated

by NOX2 play a critical role in the induction of inflam-

matory responses and tissue injury in sepsis.

The family of Toll-like receptors (TLR), which to date

contains ten members, recognizes specific molecules con-

served among microorganisms and pathogens, and plays an

important role in initiating the inflammatory response [79].

Emerging evidence demonstrates that NOX enzymes

modulate Toll-like receptor 4 (TLR4) and TLR2 signaling

not only in neutrophils and macrophages but also in other

cells. Lipopolysaccharide specifically binds to LPS-binding

protein (LBP) and forms a complex that activates the TLR4

receptor of macrophages and others cells. This interaction

triggers the activation of Ijb kinase and the mitogen-acti-

vated protein kinase kinases (MAPKK), which in turn

activate NF-jB and AP1, respectively [80]. Activated

NF-jB and AP-1 translocate into the nucleus where they

bind to DNA promoter regions and induce the transcription

NOX enzymes and acute lung injury 2377

123

Page 6: NOX enzymes: potential target for the treatment of acute lung injury

of inflammatory genes. It has been shown that in neutrophils,

NOX2-derived ROS regulate the TLR4-mediated activa-

tion of NF-jB. In addition, in endothelial cells, NOX2 also

contributes to TLR2 gene activation in response to LPS

[36, 81]. A recent study demonstrated that ROS generated

by NOX2 in neutrophils mediates high mobility group box

1 (HMGB1)/TLR4 signaling and tissue damage after

hemorrhagic shock/resuscitation in mice [82]. Besides

NOX2, NOX4 is able to directly interact with TLR4 and

mediate ROS generation and NF-jB activation in

HEK293T cells [83]. More recently, NOX4 has been

shown to mediate LPS-induced NK-jB-dependent IL8,

MCP-1 and ICAM-1 gene expression in human aortic

endothelial cells [84] and interferon regulatory factor

(IRF)-3 transcription factor activation in U373/CD14 cells

[85]. NADPH oxidase4 is also able to activate AP-1 and

subsequent CXCR6 expression, after LPS challenge in

human aortic smooth muscle cells [86]. Similar to NOX4,

the presence of crosstalk between NOX1 and TLR4 was

suggested by the observation that LPS derived from Heli-

cobacter pylori increases ROS production and NOX1

expression in guinea pig gastric pit cells through a TLR4

signaling pathway [87]. All these results suggest that NOX

family plays an important role in the activation of TLR4

signaling pathways (including NF-jB, IRF-3 and AP1) in

response to LPS. Nevertheless, the molecular mechanism

linking TLR4 to NOX1 remains unclear and the function of

NOX1 and NOX4 in TLR-mediated signaling in vivo need

to be elucidated using either knock-out mice or RNA

interference strategy.

Endothelial and epithelial targets

Alveolar cell death has been reported extensively in

humans and in experimental models of acute lung injury

[88]. Indeed, in the acute phase, the presence of edema in

the air spaces and hyaline membrane deposits are direct

consequences of alveolar-capillary barrier damage. Epi-

thelial type II cells after being initially injured, often by an

unknown stimulus, proliferate in order to repair the dam-

aged epithelium [89]. To date, it is not known whether the

alveolar-capillary barrier integrity depends preferentially

on the endothelial or the epithelial side in acute lung

damage and which are the signaling pathways involved in

alveolar cell death. Nevertheless, there is evidence that

both epithelial and endothelial cells are damaged by ROS-

dependent mechanisms and in particular by NOX-depen-

dent ROS generation.

Endothelial cell target

The injury of endothelial cells is mostly studied in sepsis-

like models using systemic injection of LPS or TNF-a.

During ALI, the endothelium undergoes large transforma-

tions in terms of expression of adhesion molecules, tight

junctions, and ROS-producing enzymes [90]. In this con-

text, NOX-derived ROS participates in the damage of

endothelial cells either by the direct activation of endo-

thelial signaling or via neutrophils or macrophages.

Lipopolysaccharide can directly increase ROS genera-

tion through the modulation of NOX enzymes in

endothelial cells [91]. A recent study demonstrates that in

these cells, stimulation by LPS leads to the activation of

IL8, which in turn regulates the expression and the activity

of NOX1 and contributes to the progression of the sepsis

cascade. These data suggest that LPS/IL8 signaling is

NOX1-dependent in endothelial cells [92]. In addition to

NOX1, NOX4, which is also expressed in endothelial cells,

is responsible for LPS/TLR4-induced ROS generation and

gene expression of chemokines such as IL8, MCP-1, and

intracellular adhesion molecule-1 (ICAM-1) in human

aortic endothelial cells [84]. The authors also demonstrated

that the specific inhibition of NOX4, by siRNA strategy,

contributes to the decrease of LPS-induced migration and

adhesion of monocytes to endothelial cells [84].

Thus, besides the direct effect of LPS on NOX activa-

tion in endothelial cells, excessive production of ROS by

NOX enzymes located in inflammatory cells has been

associated to endothelial cell damage in sepsis. Fan et al.

[34, 81] and others demonstrated that in endothelial cells,

LPS/TLR4-induced NF-jB and TLR2 gene activation is

dependent on NOX2 located in neutrophils. In addition,

neutrophilic NOX2 contributes to TNF-a-induced NF-jB-

dependent lung inflammation and endothelial cell injury in

mice [38] and participates in the activation of NF-jB and

the induction of TLR2 in endothelial cells [36]. More

recently, Farley et al. [41] reported that co-culture of

p47phox and gp91phox-deficient macrophages with pul-

monary microvascular endothelial cells stimulated with a

mix of cytokines such as TNF-a, IL1-b, and IFN-c led to a

significant decrease in endothelial cell injury, supporting

the concept that ROS produced by phagocytic NOX2 play

a crucial role in the injury of endothelial cells.

Epithelial cell target

Although the epithelial barrier after being injured by an

unknown stimulus is mostly able to repair, the persistence

of a lung injury leads to the development of fibrosis. It is

considered that epithelial cell death is crucial not only in

the weakening of the alveolar-capillary barrier, but also in

lung abnormal repair, which leads to pulmonary fibrosis

[93]. While the pathogenesis of pulmonary fibrosis, a lethal

lung disorder characterized by abnormal lung repair, is

unknown, it involves early inflammatory steps and late

fibrotic changes with proliferation of fibroblasts and their

2378 S. Carnesecchi et al.

123

Page 7: NOX enzymes: potential target for the treatment of acute lung injury

differentiation into myofibroblasts [94, 95]. Intratracheal

instillation of bleomycin in mice, a well-known charac-

terized model to study initial lung epithelial injury and

subsequent fibrosis, mimics ALI/ARDS features occurring

during the late proliferative and fibrotic phase. In this

model, NOX-dependent ROS are not only responsible for

initial epithelial damage but also for the differentiation of

fibroblasts into myofibroblasts, the hallmark of the disease.

Several studies have shown that TGF-b1 increases ROS

levels by up-regulating NOX4 expression in lung fibro-

blasts and induces their differentiation into myofibroblasts

[8, 96, 97]. Interestingly, in human idiopathic pulmonary

fibrosis (IPF), NOX4 has been detected in myofibroblasts

of late fibrotic scars, suggesting a possible role of NOX4 in

the development of organized fibrosis, and was also

detected in the alveolar proliferative epithelium of IPF

lungs adjacent to fibroblastic foci. In mice, we demon-

strated that NOX4 deficiency as well as acute treatment

with NOX inhibitors blunted TGF-b1-induced alveolar

epithelial cell death and prevented subsequent pulmonary

fibrosis [7].

The role of NOX2 and NOX1 in bleomycin-induced

lung fibrosis has also been investigated in NOX2 and

NOX1-deficient mice. Only a moderate protection from

bleomycin-induced lung fibrosis was observed in NOX2-

deficient mice [98]; however, extrapolation to human IPF is

difficult as inflammation might not be as prominent in

humans compared to mice. We found that NOX1-deficient

mice were not protected from bleomycin-induced fibrosis

(personal data). Finally, NOX4 rather than NOX1 and

NOX2 could be a good candidate for the treatment of

ARDS/ALI patients during both the acute and the prolif-

erative stage.

Epithelium and endothelium targets

Aspiration of the gastric content is considered to be an

important cause of ALI/ARDS. In addition to the low pH,

the gastric content contains particulate bacterial material,

which contributes to lung injury [99]. The intratracheal

instillation of hypochlorite (HCl) is a well-used model for

inducing lung injury secondary to gastric acid aspiration in

mice. Aspiration-induced lung injury, which depends on

neutrophilic influx into the alveolar space, is characterized

by the damage of both epithelial and endothelial cells

leading to alveolar hemorrhage and edema. Some studies

have implicated ROS as a key element in the pathogenesis

of ALI/ARDS following gastric content aspiration in

mouse models [100], but to date, only one has demon-

strated the role of NOX enzyme in this context. Indeed,

exposure of p47phox-deficient mice to HCl leads to

increased pulmonary neutrophilic infiltration, alveolar-

capillary barrier leakage, and enhanced level of pro-

inflammatory cytokine compared to WT mice [101], sug-

gesting a protective role of NOX2 in HCl-induced lung

injury by modulating the inflammatory response.

Mechanical ventilation (MV) is the unique strategy used

in patients with acute hypoxemic respiratory failure to

improve arterial oxygenation and their survival [102].

However, this therapy provokes tissue injury due to

mechanical stretch (MS). Mechanical ventilation associ-

ated with alveolar barrier overstretching contributes to

neutrophilic infiltration, release of pro-inflammatory cyto-

kines, and lung injury [103]. The cellular mechanisms

involved in MV-induced lung injury (MVILI) and

-inflammation remain unknown. A high level of ROS is

thought to be one potential initiating signal in response to

MV following mechanical stress. Indeed, treatment with

N-acetyl cysteine (NAC) attenuates MV-induced neuro-

philic influx into alveolar spaces and reduces epithelial cell

apoptosis in rats [104, 105]. Besides mitochondrial

enzymes, NOXes have been shown to contribute to ROS

production in response to mechanical stress in different

cells such as endothelial cells, epithelial cells, and vascular

smooth muscle cells [106–111]. It has been described that

NOX activation was associated with a membrane translo-

cation of p47phox in smooth muscle cells (SMC) [108, 109].

On the other hand, exposure of vascular SMCs to MV

leads to p47phox membrane translocation followed by an

increased NOX1 mRNA expression and ROS production

[108], suggesting a role for NOX1 in MVILI. Some studies

also demonstrated that ROS produced by NOX enzymes

participated in cyclic stretch-induced vascular remodeling

in SMC via matrix metalloproteinase-2 activation [108].

The NOX isoform involved in MV-induced lung injury and

the NOX-dependent signal transduction pathways need to

be clarified.

Hyperoxia-induced acute lung injury is one of the most

relevant models of oxidative stress and alveolar cell death,

which is not closely linked to the magnitude of the

inflammatory response. In rodents and in alveolar cell

culture, oxygen toxicity (100 % O2) has been used as a

well-established model of lesions mimicking the acute

phase of ALI/ARDS and for studying direct alveolar

damage induced by high levels of oxidants. It was first

explored in rats and later extensively characterized in mice

[112–114]. During the initiation phase (usually lasting for

48 h), only subtle changes can be detected, such as the

arrest of cell replication, and lesions are not evident on

light microscopy. This phase is followed by diffuse alve-

olar damage with hyaline membrane deposition and

extensive death of alveolar cells (mainly endothelial and

epithelial cells) associated with a generally mild inflam-

matory response, which can vary according to the species

[115]. Alveolar cell death has been shown to be directly

related to increased generation of oxidant in hyperoxic

NOX enzymes and acute lung injury 2379

123

Page 8: NOX enzymes: potential target for the treatment of acute lung injury

condition [116]. Reactive oxygen species can be generated

by mitochondrial chain transport as well as by NADPH

oxidase enzymes [117]. In vitro studies have shown that the

diphenyleneiodonium (DPI), a non-specific inhibitor of

NOX isoforms, was effective in reducing hyperoxia-

induced ROS generation in a pulmonary epithelial cell line

(MLE-12) and in primary pulmonary type II cells [117–

119]. Recently, our laboratory demonstrated that NOX1,

which is highly expressed in lungs, plays a crucial role in

hyperoxia-induced acute lung injury [17]. NADPH oxi-

dase1-deficient mice exposed to hyperoxia exhibited

reduced pulmonary edema, hyaline membrane deposition,

and alveolar-capillary damage. Indeed, in situ lung cell

death was markedly decreased in NOX1-deficient mice and

paralleled with decreased ROS production and cell death in

endothelial and epithelial cells. The phosphorylation of

both c-Jun N-terminal kinase (JNK) and extracellular sig-

nal-regulated kinase (ERK), as well as caspase-3 activation

were decreased in lung homogenates. All these results

demonstrate a role for NOX1 in hyperoxia-mediated bar-

rier dysfunction; however, the question of the contribution

of NOX1 restricted to the epithelial side or to endothelial

side or both in ALI/ARDS and its precise cellular signaling

pathway is still open.

As stated above, the integrity of the alveolar-capillary

barrier depends not only on the epithelium but also on the

endothelium. Recent studies have shown increased NOX1

mRNA expression in mouse lung endothelial cells after

hyperoxic condition [17, 28] as well as NOX1 contribution

to endothelial cell death [17]. The involvement of NOX4 in

hyperoxic cultured endothelial cells (EC) has also been

investigated. NADPH oxidase4 mRNA expression is

increased in hyperoxia [28] through direct modulation of

gene transcription. Indeed, direct interaction between nrf2

transcription factor and NOX4 promoter has been reported.

The authors demonstrated that hyperoxia increased the

recruitment- and the binding of nfr2 to endogenous NOX4

promoter via antioxidant response element (ARE) in pul-

monary endothelial cells [44]. Hyperoxia also regulates

NOX activation in part by ERK1/2 and p38 MAPK [117],

but also by an Src-dependent tyrosine phosphorylation of

p47phox [29]. It has also been proposed that NOX2 could be

activated by tyrosine phosphorylation of cortactin and

p47phox translocation following hyperoxia in ECs derived

from human pulmonary artery [120]. Recently, Pendyala

et al. [28] demonstrated the contribution of NOX4 in

endothelial dysfunction. They showed that the transfection

of a NOX4-specific siRNA in HPAEC attenuated hyper-

oxia-induced migration and capillary tube formation.

Therefore, we have now convincing evidence of NOXs

activation (NOX1, 2 and 4) by oxidative stress in murine

and human endothelial cells [17, 29, 117, 120, 121].

Although we have less evidence, we could also hypothesize

that NOX4 participates in epithelial cell damage in ALI/

ARDS. Indeed, we recently reported that NOX4 mRNA

was expressed in primary type II epithelial cells and

mediated TGF-b1-induced epithelial cell death [7]. These

results suggest that NOX4 contributes not only to the

dysfunction or death of endothelial cells, but it might be

involved in the alveolar-capillary disruption observed in

ALI/ARDS. However, all these experiments were per-

formed essentially in cell cultures and should be confirmed

first by using NOX4-deficient mice or/and transfection of

NOX4-specific siRNA and then in humans.

Controversial studies concerning the role of NOX2 in

hyperoxia-induced lung injury have been reported.

Pendyala showed that NOX2-deficient mice exposed to

acute hyperoxia developed attenuated pulmonary edema,

lung fibrosis, and weak inflammatory response [122]. By

contrast, we found that NOX2-deficient mice exposed to

hyperoxia were not protected and display a huge neutrophil

influx in BAL, alveolar cell death, and lung injury [17],

suggesting that NOX2 does not mediate alveolar-capillary

disruption in hyperoxia. Indeed, neutrophil or macrophage

depletion did not change lung damage in hyperoxic lung

injury [123, 124]. Similarly, NOX2-deficient mice exposed

to 48 h of hyperoxia following acid aspiration showed a

greater amount of neutrophils compared to WT mice,

without modification of lung injury [73].

NOX as treatment of ALI/ARDS

As largely described above, NOX inhibitors might have

potential in vivo use in ARDS. However, the multiplicity

of lung cells combined with the cellular and functional

specificity of the different NOX isoforms makes this

approach delicate. Moreover, the measurement of NOX

activity is often indirect since it is evaluated by the dosage

of ROS-derived products using colorimetric or fluorescent

probes. One must be aware that measuring ROS or derived

products levels might not only be due to ROS production

by NOX enzymes but also by other ROS-producing

enzymes. Therefore the specific efficacy of NOX inhibition

can be difficult to prove.

Today, therapeutic intervention for ALI/ARDS consists

of protecting the lung by using adaptive mechanical ven-

tilation and oxygenation and thus limiting mortality [125].

Lung-protective mechanical ventilation with lower tidal

volumes in patients not suffering from acute lung injury: a

review of clinical studies. This strategy was elaborated

according to the results obtained in clinical trials and in

experimental animal models [126]. Some studies have

suggested that subgroups of patients may benefit from

targeted therapeutic interventions. Most promising is the

differentiation between patients in early versus late-phase

2380 S. Carnesecchi et al.

123

Page 9: NOX enzymes: potential target for the treatment of acute lung injury

ARDS, direct versus indirect lung injury, and patients with

altered coagulation. A high dose of corticosteroid admin-

istration did not improve mortality, whereas low to

moderate doses appear to be harmful if initiated later and

are of unclear benefit [127, 128]. Surfactant supplementa-

tion was shown to be helpful only in pediatric patients with

direct lung injury [129] and anticoagulants may be suc-

cessful in the subgroup of patients with vascular disease

[130]. There is an interest in developing NOX inhibitors,

since they can act for some of them in the early phase and

for other in the fibro-proliferative phase. Table 2 summa-

rizes the expected effects of the potential NOX inhibitors

for the treatment of ARDS.

NADPH oxidase1 inhibition might be useful in the acute

phase of ALI/ARDS since it interferes with endothelial and

epithelial cell death either by decreasing oxidative stress-

induced genotoxicity or by affecting MAPK signaling

pathways [17]. However, this study was performed only in

mice and evidence in humans is still required. The possi-

bility that NOX1 inhibition can affect TNF activation

might also be interesting in case of ARDS due to sepsis

[22].

NADPH oxidase2 inhibition effects are more complex

and the results are somehow controversial. NADPH oxi-

dase2 is present mainly in phagocytic cells, but also in a

great amount in endothelial cells. Indeed, changing

phagocyte killing might be dangerous in situations of

ARDS due to sepsis or to unknown origin even if ROS-

produced by NOX2 in other cells might prevent concom-

itantly. In this case, a tagged-cell inhibitor would be ideal,

but at the present time rather difficult.

More promising would be the inhibition of NOX4,

which is present in several lung cells epithelial and fibro-

blasts. Several studies have shown a very robust effect in

decreasing epithelial cell death initiated by TGF-b1 and

myofibroblast differentiation [7, 8]. Moreover, NOX4

action is upstream of the pleiomorph effects of TGF-b1,

and therefore could be more efficient in blocking the TGF-

b1 deleterious cascade. We can also hypothesize, as NOX4

is strongly expressed in epithelial cells and its signaling

potentiates cell death induced by TGF-b1, that NOX4-

specific inhibition could prevent the alveolar-capillary

disruption in the ARDS early phase [7]. NADPH oxidase4

is also involved in TLR4 signaling mediated by LPS and

might therefore participate in endothelial dysfunction [84].

This study has been performed in vitro, and more in vivo

data are needed before envisaging therapeutic possibilities.

Conclusions

NADPH oxidase enzymes, which are widely expressed in

different lung cell types, participate not only in the main-

tenance of physiological processes in lungs but also

contribute to the pathogenesis of acute lung diseases such

as ALI and ARDS. The multiplicity of the well-charac-

terized animal models mimicking ARDS/ALI, the use of

NOX-deficient mice and in vivo siRNA transfection strat-

egies allowed to explore NOX-dependent cellular and

molecular mechanisms involved in the development of the

disease and finally envisage new therapeutic approaches.

Developing NOX inhibitors could therefore be a promising

treatment concept for ARDS/ALI. Nevertheless, at the

present time, no direct method for measuring specifically

NOX-dependent ROS generation has been developed to

prove the efficacy of NOX inhibitors and specific inhibitors

for one single NOX isoform are not available. Whether in

some case it might be useful to target two different iso-

forms concomitantly, such as in early phase, in other

situations such as ARDS induced by sepsis, this could be

deleterious due to combined unwarranted secondary

effects. Thus, further in vivo studies concerning NOX

inhibitors are necessary to prove their clinical utility in the

management of ALI/ARDS.

Table 2 Summary of potential effect of NOX inhibitors in ALI/ARDS

NOX isoform

inhibitors

ARDS/ALI clinical stages Target cells Expected effects Secondary effects

NOX1 Acute stage: alveolar-

capillary barrier disruption

Epithelial and

endothelial cells

Decreased cell death (genotoxicity, MAPK signaling,

TNF-RI-JNK signaling)

N.D

NOX2 Acute stage: inflammation/

endothelial cell injury

Macrophages/

neutrophils

Decreased inflammatory response, endothelial cell

death, crosstalk with TLR4 signaling

Increased

susceptibility to

infection

Endothelial cells Decreased cell death, crosstalk with TLR2 signaling N.D

NOX4 Acute stage: alveolar-

capillary barrier disruption

Epithelial cells Decreased cell death (genotoxicity) interference with

TGF-b signaling

N.D

Acute stage: inflammation/

endothelial cell injury

Endothelial cells Decreased cell death,TLR4 crosstalk signaling N.D

Fibro-proliferative stage Myofibroblasts Decreased proliferation and differentiation,

interference with TGF-b1 signaling

N.D

NOX enzymes and acute lung injury 2381

123

Page 10: NOX enzymes: potential target for the treatment of acute lung injury

References

1. Tasaka S, Amaya F, Hashimoto S, Ishizaka A (2008) Roles of

oxidants and redox signaling in the pathogenesis of acute

respiratory distress syndrome. Antioxid Redox Signal

10:739–753

2. Bedard K, Krause KH (2007) The NOX family of ROS-gener-

ating NADPH oxidases: physiology and pathophysiology.

Physiol Rev 87:245–313

3. Chabot F, Mitchell JA, Gutteridge JM, Evans TW (1998)

Reactive oxygen species in acute lung injury. Eur Respir J

11:745–757

4. Lambeth JD (2004) NOX enzymes and the biology of reactive

oxygen. Nat Rev Immunol 4:181–189

5. Griffith B, Pendyala S, Hecker L, Lee PJ, Natarajan V, Than-

nickal VJ (2009) NOX enzymes and pulmonary disease.

Antioxid Redox Signal 11:2505–2516

6. van der Vliet A (2008) NADPH oxidases in lung biology and

pathology: host defense enzymes, and more. Free Radic Biol

Med 44:938–955

7. Carnesecchi S, Deffert C, Donati Y et al (2011) A key role for

NOX4 in epithelial cell death during development of lung

fibrosis. Antioxid Redox Signal 15(3):607–619

8. Hecker L, Vittal R, Jones T et al (2009) NADPH oxidase-4

mediates myofibroblast activation and fibrogenic responses to

lung injury. Nat Med 15:1077–1081

9. Sorescu D, Weiss D, Lassegue B et al (2002) Superoxide pro-

duction and expression of nox family proteins in human

atherosclerosis. Circulation 105:1429–1435

10. Zhang X, Shan P, Jiang G, Cohn L, Lee PJ (2006) Toll-like

receptor 4 deficiency causes pulmonary emphysema. J Clin

Invest 116:3050–3059

11. Rada B, Leto TL (2008) Oxidative innate immune defenses by

Nox/Duox family NADPH oxidases. Contrib Microbiol

15:164–187

12. Thannickal VJ, Fanburg BL (2000) Reactive oxygen species in

cell signaling. Am J Physiol Lung Cell Mol Physiol 279:L1005–

L1028

13. Cheng G, Cao Z, Xu X, van Meir EG, Lambeth JD (2001)

Homologs of gp91phox: cloning and tissue expression of Nox3,

Nox4, and Nox5. Gene 269:131–140

14. Kikuchi H, Hikage M, Miyashita H, Fukumoto M (2000)

NADPH oxidase subunit, gp91(phox) homologue, preferentially

expressed in human colon epithelial cells. Gene 254:237–243

15. Geiszt M, Witta J, Baffi J, Lekstrom K, Leto TL (2003) Dual

oxidases represent novel hydrogen peroxide sources supporting

mucosal surface host defense. Faseb J 17:1502–1504

16. Edens WA, Sharling L, Cheng G et al (2001) Tyrosine cross-

linking of extracellular matrix is catalyzed by Duox, a multi-

domain oxidase/peroxidase with homology to the phagocyte

oxidase subunit gp91phox. J Cell Biol 154:879–891

17. Carnesecchi S, Deffert C, Pagano A et al (2009) NADPH oxi-

dase-1 plays a crucial role in hyperoxia-induced acute lung

injury in mice. Am J Respir Crit Care Med 180:972–981

18. Mittal M, Roth M, Konig P et al (2007) Hypoxia-dependent

regulation of nonphagocytic NADPH oxidase subunit NOX4 in

the pulmonary vasculature. Circ Res 101:258–267

19. Harper RW, Xu C, Eiserich JP et al (2005) Differential regu-

lation of dual NADPH oxidases/peroxidases, Duox1 and Duox2,

by Th1 and Th2 cytokines in respiratory tract epithelium. FEBS

Lett 579:4911–4917

20. Nagai K, Betsuyaku T, Suzuki M et al (2008) Dual oxidase 1

and 2 expression in airway epithelium of smokers and patients

with mild/moderate chronic obstructive pulmonary disease.

Antioxid Redox Signal 10:705–714

21. Garrido-Urbani S, Jemelin S, Deffert C et al (2011) Correction:

targeting vascular NADPH oxidase 1 blocks tumor angiogenesis

through a PPARalpha mediated mechanism. PLoS One 6(2):

e14665

22. Pantano C, Anathy V, Ranjan P, Heintz NH, Janssen-Heininger

YM (2007) Nonphagocytic oxidase 1 causes death in lung epi-

thelial cells via a TNF-RI-JNK signaling axis. Am J Respir Cell

Mol Biol 36:473–479

23. Goyal P, Weissmann N, Grimminger F et al (2004) Upregulation

of NAD(P)H oxidase 1 in hypoxia activates hypoxia-inducible

factor 1 via increase in reactive oxygen species. Free Radic Biol

Med 36:1279–1288

24. Malec V, Gottschald OR, Li S, Rose F, Seeger W, Hanze J

(2010) HIF-1 alpha signaling is augmented during intermittent

hypoxia by induction of the Nrf2 pathway in NOX1-expressing

adenocarcinoma A549 cells. Free Radic Biol Med 48:1626–

1635

25. Puca R, Nardinocchi L, Starace G et al (2010) Nox1 is involved

in p53 deacetylation and suppression of its transcriptional

activity and apoptosis. Free Radic Biol Med 48:1338–1346

26. Ranjan P, Anathy V, Burch PM, Weirather K, Lambeth JD,

Heintz NH (2006) Redox-dependent expression of cyclin D1

and cell proliferation by Nox1 in mouse lung epithelial cells.

Antioxid Redox Signal 8:1447–1459

27. Clempus RE, Sorescu D, Dikalova AE et al (2007) Nox4 is

required for maintenance of the differentiated vascular smooth

muscle cell phenotype. Arterioscler Thromb Vasc Biol 27:42–48

28. Pendyala S, Gorshkova IA, Usatyuk PV et al (2009) Role of

Nox4 and Nox2 in hyperoxia-induced reactive oxygen species

generation and migration of human lung endothelial cells.

Antioxid Redox Signal 11:747–764

29. Chowdhury AK, Watkins T, Parinandi NL et al (2005) Src-

mediated tyrosine phosphorylation of p47phox in hyperoxia-

induced activation of NADPH oxidase and generation of

reactive oxygen species in lung endothelial cells. J Biol Chem

280:20700–20711

30. Fu XW, Wang D, Nurse CA, Dinauer MC, Cutz E (2000)

NADPH oxidase is an O2 sensor in airway chemoreceptors:

evidence from K? current modulation in wild-type and oxidase-

deficient mice. Proc Natl Acad Sci USA 97:4374–4379

31. Al-Mehdi AB, Zhao G, Dodia C et al (1998) Endothelial

NADPH oxidase as the source of oxidants in lungs exposed to

ischemia or high K?. Circ Res 83:730–737

32. Liu JQ, Erbynn EM, Folz RJ (2005) Chronic hypoxia-enhanced

murine pulmonary vasoconstriction: role of superoxide and

gp91phox. Chest 128:594S–596S

33. Liu JQ, Zelko IN, Erbynn EM, Sham JS, Folz RJ (2006) Hyp-

oxic pulmonary hypertension: role of superoxide and NADPH

oxidase (gp91phox). Am J Physiol Lung Cell Mol Physiol

290:L2–L10

34. Fan J, Frey RS, Malik AB (2003) TLR4 signaling induces TLR2

expression in endothelial cells via neutrophil NADPH oxidase.

J Clin Invest 112:1234–1243

35. Cutz E, Pan J, Yeger H (2009) The role of NOX2 and ‘‘novel

oxidases’’ in airway chemoreceptor O(2) sensing. Adv Exp Med

Biol 648:427–438

36. Fan J, Frey RS, Rahman A, Malik AB (2002) Role of neutrophil

NADPH oxidase in the mechanism of tumor necrosis factor-

alpha induced NF-kappa B activation and intercellular adhesion

molecule-1 expression in endothelial cells. J Biol Chem

277:3404–3411

37. Kong X, Thimmulappa R, Kombairaju P, Biswal S (2010)

NADPH oxidase-dependent reactive oxygen species mediate

amplified TLR4 signaling and sepsis-induced mortality in Nrf2-

deficient mice. J Immunol 185:569–577

2382 S. Carnesecchi et al.

123

Page 11: NOX enzymes: potential target for the treatment of acute lung injury

38. Zhang WJ, Wei H, Tien YT, Frei B (2011) Genetic ablation of

phagocytic NADPH oxidase in mice limits TNFalpha-induced

inflammation in the lungs but not other tissues. Free Radic Biol

Med 50:1517–1525

39. Vlahos R, Stambas J, Bozinovski S, Broughton BR, Drummond

GR, Selemidis S (2011) Inhibition of Nox2 oxidase activity

ameliorates influenza A virus-induced lung inflammation. PLoS

Pathog 7:e1001271

40. Soucy-Faulkner A, Mukawera E, Fink K et al (2010) Require-

ment of NOX2 and reactive oxygen species for efficient RIG-I-

mediated antiviral response through regulation of MAVS

expression. PLoS Pathog 6:e1000930

41. Farley KS, Wang L, Mehta S (2009) Septic pulmonary micro-

vascular endothelial cell injury: role of alveolar macrophage

NADPH oxidase. Am J Physiol Lung Cell Mol Physiol

296:L480–L488

42. Kampfrath T, Maiseyeu A, Ying Z et al (2011) Chronic fine

particulate matter exposure induces systemic vascular dysfunc-

tion via NADPH oxidase and TLR4 pathways. Circ Res

108:716–726

43. Lane CR, Lee PJ (2010) NOX3 regulates the inflammatory

response and endothelial barrier integrity in hyperoxic acute

lung injury. Am J respir Crit Care Med 181:A2719

44. Pendyala S, Moitra J, Kalari S et al (2011) Nrf2 regulates

hyperoxia-induced Nox4 expression in human lung endothe-

lium: identification of functional antioxidant response elements

on the Nox4 promoter. Free Radic Biol Med 50:1749–1759

45. Amara N, Bachoual R, Desmard M et al (2007) Diesel exhaust

particles induce matrix metalloprotease-1 in human lung epithe-

lial cells via a NADP(H) oxidase/NOX4 redox-dependent

mechanism. Am J Physiol Lung Cell Mol Physiol 293:L170–L181

46. Pache JC, Carnesecchi S, Deffert C et al (2011) NOX-4 is

expressed in thickened pulmonary arteries in idiopathic pul-

monary fibrosis. Nat Med 17:31–32 author reply 2–3

47. Sturrock A, Cahill B, Norman K et al (2006) Transforming

growth factor-beta1 induces Nox4 NAD(P)H oxidase and reac-

tive oxygen species-dependent proliferation in human

pulmonary artery smooth muscle cells. Am J Physiol Lung Cell

Mol Physiol 290:L661–L673

48. Park S, Ahn JY, Lim MJ et al (2010) Sustained expression of

NADPH oxidase 4 by p38 MAPK-Akt signaling potentiates

radiation-induced differentiation of lung fibroblasts. J Mol Med

88:807–816

49. Dhaunsi GS, Paintlia MK, Kaur J, Turner RB (2004) NADPH

oxidase in human lung fibroblasts. J Biomed Sci 11:617–622

50. Li S, Tabar SS, Malec V et al (2008) NOX4 regulates ROS

levels under normoxic and hypoxic conditions, triggers prolif-

eration, and inhibits apoptosis in pulmonary artery adventitial

fibroblasts. Antioxid Redox Signal 10:1687–1698

51. Rada B, Leto TL (2010) Characterization of hydrogen peroxide

production by Duox in bronchial epithelial cells exposed to

Pseudomonas aeruginosa. FEBS Lett 584:917–922

52. Boots AW, Hristova M, Kasahara DI, Haenen GR, Bast A, van

der Vliet A (2009) ATP-mediated activation of the NADPH

oxidase DUOX1 mediates airway epithelial responses to bac-

terial stimuli. J Biol Chem 284:17858–17867

53. Lavigne MC, Eppihimer MJ (2005) Cigarette smoke condensate

induces MMP-12 gene expression in airway-like epithelia.

Biochem Biophys Res Commun 330:194–203

54. Nakanaga T, Nadel JA, Ueki IF, Koff JL, Shao MX (2007)

Regulation of interleukin-8 via an airway epithelial signaling

cascade. Am J Physiol Lung Cell Mol Physiol 292:L1289–

L1296

55. Shao MX, Nadel JA (2005) Dual oxidase 1-dependent

MUC5AC mucin expression in cultured human airway epithelial

cells. Proc Natl Acad Sci USA 102:767–772

56. Wesley UV, Bove PF, Hristova M, McCarthy S, van der Vliet A

(2007) Airway epithelial cell migration and wound repair by

ATP-mediated activation of dual oxidase 1. J Biol Chem

282:3213–3220

57. Schwarzer C, Machen TE, Illek B, Fischer H (2004) NADPH

oxidase-dependent acid production in airway epithelial cells.

J Biol Chem 279:36454–36461

58. Koff JL, Shao MX, Kim S, Ueki IF, Nadel JA (2006) Pseudo-

monas lipopolysaccharide accelerates wound repair via

activation of a novel epithelial cell signaling cascade. J Immunol

177:8693–8700

59. Fischer H, Gonzales LK, Kolla V et al (2007) Developmental

regulation of DUOX1 expression and function in human fetal

lung epithelial cells. Am J Physiol Lung Cell Mol Physiol

292:L1506–L1514

60. Forteza R, Salathe M, Miot F, Conner GE (2005) Regulated

hydrogen peroxide production by Duox in human airway epi-

thelial cells. Am J Respir Cell Mol Biol 32:462–469

61. Ware LB, Matthay MA (2000) The acute respiratory distress

syndrome. N Engl J Med 342:1334–1349

62. Bachofen M, Weibel ER (1977) Alterations of the gas exchange

apparatus in adult respiratory insufficiency associated with

septicemia. Am Rev Respir Dis 116:589–615

63. Pache JC, Christakos PG, Gannon DE, Mitchell JJ, Low RB,

Leslie KO (1998) Myofibroblasts in diffuse alveolar damage of

the lung. Mod Pathol 11:1064–1070

64. Bachofen M, Weibel ER (1982) Structural alterations of lung

parenchyma in the adult respiratory distress syndrome. Clin

Chest Med 3:35–56

65. Prescott SM, McIntyre TM, Zimmerman G (1999) Two of the

usual suspects, platelet-activating factor and its receptor,

implicated in acute lung injury. J Clin Invest 104:1019–1020

66. Matthay MA, Geiser T, Matalon S, Ischiropoulos H (1999)

Oxidant-mediated lung injury in the acute respiratory distress

syndrome. Crit Care Med 27:2028–2030

67. Matute-Bello G, Frevert CW, Martin TR (2008) Animal models

of acute lung injury. Am J Physiol Lung Cell Mol Physiol

295:L379–L399

68. Salvemini D, Cuzzocrea S (2002) Oxidative stress in septic

shock and disseminated intravascular coagulation. Free Radic

Biol Med 33:1173–1185

69. Victor VM, Rocha M, De la Fuente M (2003) Regulation of

macrophage function by the antioxidant N-acetylcysteine in

mouse-oxidative stress by endotoxin. Int Immunopharmacol

3:97–106

70. Ben-Shaul V, Lomnitski L, Nyska A, Zurovsky Y, Bergman M,

Grossman S (2001) The effect of natural antioxidants, NAO and

apocynin, on oxidative stress in the rat heart following LPS

challenge. Toxicol Lett 123:1–10

71. Koay MA, Christman JW, Segal BH et al (2001) Impaired pul-

monary NF-kappaB activation in response to lipopolysaccharide

in NADPH oxidase-deficient mice. Infect Immun 69:5991–5996

72. Forman HJ, Torres M (2002) Reactive oxygen species and cell

signaling: respiratory burst in macrophage signaling. Am J

Respir Crit Care Med 166:S4–S8

73. Gao XP, Standiford TJ, Rahman A et al (2002) Role of NADPH

oxidase in the mechanism of lung neutrophil sequestration and

microvessel injury induced by Gram-negative sepsis: studies in

p47phox-/- and gp91phox-/- mice. J Immunol 168:3974–3982

74. Sadikot RT, Zeng H, Yull FE et al (2004) p47phox deficiency

impairs NF-kappa B activation and host defense in Pseudomo-nas pneumonia. J Immunol 172:1801–1808

75. Sato K, Kadiiska MB, Ghio AJ et al (2002) In vivo lipid-derived

free radical formation by NADPH oxidase in acute lung injury

induced by lipopolysaccharide: a model for ARDS. FASEB J

16:1713–1720

NOX enzymes and acute lung injury 2383

123

Page 12: NOX enzymes: potential target for the treatment of acute lung injury

76. Swain SD, Wright TW, Degel PM, Gigliotti F, Harmsen AG

(2004) Neither neutrophils nor reactive oxygen species con-

tribute to tissue damage during Pneumocystis pneumonia in

mice. Infect Immun 72:5722–5732

77. Zhang WJ, Wei H, Frei B (2009) Genetic deficiency of NADPH

oxidase does not diminish, but rather enhances, LPS-induced

acute inflammatory responses in vivo. Free Radic Biol Med

46:791–798

78. Yang CS, Shin DM, Kim KH et al (2009) NADPH oxidase 2

interaction with TLR2 is required for efficient innate immune

responses to mycobacteria via cathelicidin expression. J Immu-

nol 182:3696–3705

79. Takeda K, Akira S (2004) TLR signaling pathways. Semin

Immunol 16:3–9

80. Liu SF, Malik AB (2006) NF-kappa B activation as a patho-

logical mechanism of septic shock and inflammation. Am J

Physiol Lung Cell Mol Physiol 290:L622–L645

81. Pacquelet S, Johnson JL, Ellis BA et al (2007) Cross-talk

between IRAK-4 and the NADPH oxidase. Biochem J

403:451–461

82. Fan J, Cai H, Tan WS (2007) Role of the plasma membrane

ROS-generating NADPH oxidase in CD34? progenitor cells

preservation by hypoxia. J Biotechnol 130:455–462

83. Park HS, Jung HY, Park EY, Kim J, Lee WJ, Bae YS (2004)

Cutting edge: direct interaction of TLR4 with NAD(P)H oxidase

4 isozyme is essential for lipopolysaccharide-induced produc-

tion of reactive oxygen species and activation of NF-kappa B.

J Immunol 173:3589–3593

84. Park HS, Chun JN, Jung HY, Choi C, Bae YS (2006) Role of

NADPH oxidase 4 in lipopolysaccharide-induced proinflam-

matory responses by human aortic endothelial cells. Cardiovasc

Res 72:447–455

85. Chiang E, Dang O, Anderson K, Matsuzawa A, Ichijo H, David

M (2006) Cutting edge: apoptosis-regulating signal kinase 1 is

required for reactive oxygen species-mediated activation of IFN

regulatory factor 3 by lipopolysaccharide. J Immunol

176:5720–5724

86. Patel DN, Bailey SR, Gresham JK et al (2006) TLR4-NOX4-

AP-1 signaling mediates lipopolysaccharide-induced CXCR6

expression in human aortic smooth muscle cells. Biochem

Biophys Res Commun 347:1113–1120

87. Kawahara T, Kohjima M, Kuwano Y et al (2005) Helicobacter

pylori lipopolysaccharide activates Rac1 and transcription of

NADPH oxidase Nox1 and its organizer NOXO1 in guinea pig

gastric mucosal cells. Am J Physiol Cell Physiol 288:C450–C457

88. Kistler GS, Caldwell PR, Weibel ER (1967) Development of

fine structural damage to alveolar and capillary lining cells in

oxygen-poisoned rat lungs. J Cell Biol 32:605–628

89. Selman M, Pardo A (2006) Role of epithelial cells in idiopathic

pulmonary fibrosis: from innocent targets to serial killers. Proc

Am Thorac Soc 3:364–372

90. Maniatis NA, Orfanos SE (2008) The endothelium in acute lung

injury/acute respiratory distress syndrome. Curr Opin Crit Care

14:22–30

91. Al Ghouleh I, Magder S (2008) Nicotinamide adenine dinucle-

otide phosphate (reduced form) oxidase is important for LPS-

induced endothelial cell activation. Shock 29:553–559

92. Miyoshi T, Yamashita K, Arai T, Yamamoto K, Mizugishi K,

Uchiyama T (2010) The role of endothelial interleukin-8/

NADPH oxidase 1 axis in sepsis. Immunology 131:331–339

93. Thannickal VJ, Horowitz JC (2006) Evolving concepts of

apoptosis in idiopathic pulmonary fibrosis. Proc Am Thorac Soc

3:350–356

94. Gauldie J (2002) Pro: inflammatory mechanisms are a minor

component of the pathogenesis of idiopathic pulmonary fibrosis.

Am J Respir Crit Care Med 165:1205–1206

95. Horowitz JC, Thannickal VJ (2006) Epithelial-mesenchymal

interactions in pulmonary fibrosis. Semin Respir Crit Care Med

27:600–612

96. Crestani B, Besnard V, Boczkowski J (2011) Signalling path-

ways from NADPH oxidase-4 to idiopathic pulmonary fibrosis.

Int J Biochem Cell Biol 43(8):1086–1089

97. Amara N, Goven D, Prost F, Muloway R, Crestani B, Bocz-

kowski J (2010) NOX4/NADPH oxidase expression is increased

in pulmonary fibroblasts from patients with idiopathic pulmon-

ary fibrosis and mediates TGFbeta1-induced fibroblast

differentiation into myofibroblasts. Thorax 65:733–738

98. Manoury B, Nenan S, Leclerc O et al (2005) The absence of

reactive oxygen species production protects mice against bleo-

mycin-induced pulmonary fibrosis. Respir Res 6:11

99. Knight PR, Rutter T, Tait AR, Coleman E, Johnson K (1993)

Pathogenesis of gastric particulate lung injury: a comparison and

interaction with acidic pneumonitis. Anesth Analg 77:754–760

100. Goldman G, Welbourn R, Kobzik L, Valeri CR, Shepro D,

Hechtman HB (1992) Reactive oxygen species and elastase

mediate lung permeability after acid aspiration. J Appl Physiol

73:571–575

101. Segal BH, Davidson BA, Hutson AD et al (2007) Acid aspira-

tion-induced lung inflammation and injury are exacerbated in

NADPH oxidase-deficient mice. Am J Physiol Lung Cell Mol

Physiol 292:L760–L768

102. Girard TD, Bernard GR (2007) Mechanical ventilation in

ARDS: a state-of-the-art review. Chest 131:921–929

103. Fu Z, Costello ML, Tsukimoto K et al (1992) High lung volume

increases stress failure in pulmonary capillaries. J Appl Physiol

73:123–133

104. Syrkina O, Jafari B, Hales CA, Quinn DA (2008) Oxidant stress

mediates inflammation and apoptosis in ventilator-induced lung

injury. Respirology 13:333–340

105. Chess PR, O’Reilly MA, Sachs F, Finkelstein JN (2005) Reac-

tive oxidant and p42/44 MAP kinase signaling is necessary for

mechanical strain-induced proliferation in pulmonary epithelial

cells. J Appl Physiol 99:1226–1232

106. Chapman KE, Sinclair SE, Zhuang D, Hassid A, Desai LP,

Waters CM (2005) Cyclic mechanical strain increases reactive

oxygen species production in pulmonary epithelial cells. Am J

Physiol Lung Cell Mol Physiol 289:L834–L841

107. Matsushita H, Lee KH, Tsao PS (2001) Cyclic strain induces

reactive oxygen species production via an endothelial NAD(P)H

oxidase. J Cell Biochem Suppl Suppl 36:99–106

108. Grote K, Flach I, Luchtefeld M et al (2003) Mechanical stretch

enhances mRNA expression and proenzyme release of matrix

metalloproteinase-2 (MMP-2) via NAD(P)H oxidase-derived

reactive oxygen species. Circ Res 92:e80–e86

109. Oeckler RA, Kaminski PM, Wolin MS (2003) Stretch enhances

contraction of bovine coronary arteries via an NAD(P)H oxidase-

mediated activation of the extracellular signal-regulated kinase

mitogen-activated protein kinase cascade. Circ Res 92:23–31

110. Mata-Greenwood E, Grobe A, Kumar S, Noskina Y, Black SM(2005) Cyclic stretch increases VEGF expression in pulmonary

arterial smooth muscle cells via TGF-beta1 and reactive oxygen

species: a requirement for NAD(P)H oxidase. Am J Physiol

Lung Cell Mol Physiol 289:L288–L289

111. Black SM, Grobe A, Mata-Greenwood E, Noskina Y (2004)

Cyclic stretch increases VEGF expression in pulmonary arterial

smooth muscle cells via TGF-1 and reactive oxygen species: a

requirement for NAD(P)H oxidase. Conf Proc IEEE Eng Med

Biol Soc 7:5053–5056

112. Crapo JD, Barry BE, Foscue A, Shelburne J (1980) Structural

and biochemical changes in rat lungs occurring during exposure

to lethal and adaptive doses of oxygen. Am Rev Respir Dis

122:123–143

2384 S. Carnesecchi et al.

123

Page 13: NOX enzymes: potential target for the treatment of acute lung injury

113. Barazzone C, Horowitz S, Donati YR, Rodriguez I, Piguet PF

(1998) Oxygen toxicity in mouse lung: pathways to cell death.

Am J Respir Cell Mol Biol 19:573–581

114. Smith LJ, Friedman H, Anderson JD (1988) Hyperoxic lung

injury in mice: effect of neutrophil depletion and food depri-

vation. J Lab Clin Med 111:449–458

115. Crapo JD (1986) Morphologic changes in pulmonary oxygen

toxicity. Annu Rev Physiol 48:721–731

116. Budinger GR, Mutlu GM, Urich D et al (2010) Epithelial cell

death is an important contributor to oxidant-mediated acute lung

injury. Am J Respir Crit Care Med 183(8):1043–1054

117. Parinandi NL, Kleinberg MA, Usatyuk PV et al (2003) Hyper-

oxia-induced NAD(P)H oxidase activation and regulation by

MAP kinases in human lung endothelial cells. Am J Physiol

Lung Cell Mol Physiol 284:L26–L38

118. van Klaveren RJ, Roelant C, Boogaerts M, Demedts M, Nemery

B (1997) Involvement of an NAD(P)H oxidase-like enzyme in

superoxide anion and hydrogen peroxide generation by rat type

II cells. Thorax 52:465–471

119. Zhang X, Shan P, Sasidhar M et al (2003) Reactive oxygen

species and extracellular signal-regulated kinase 1/2 mitogen-

activated protein kinase mediate hyperoxia-induced cell death in

lung epithelium. Am J Respir Cell Mol Biol 28:305–315

120. Usatyuk PV, Romer LH, He D et al (2007) Regulation of

hyperoxia-induced NADPH oxidase activation in human lung

endothelial cells by the actin cytoskeleton and cortactin. J Biol

Chem 282:23284–23295

121. Brueckl C, Kaestle S, Kerem A et al (2006) Hyperoxia-induced

reactive oxygen species formation in pulmonary capillary

endothelial cells in situ. Am J Respir Cell Mol Biol 34:453–463

122. Natarajan V, Pendyala S, Gorshkova IA et al (2008) Role of

Nox4 and Nox2 in hyperoxia-induced reactive oxygen species

generation and migration of human lung endothelial cells.

Antioxid Redox Signal 11(4):747–764

123. Berg JT, White JE, Tsan MF (1995) Response of alveolar

macrophage-depleted rats to hyperoxia. Exp Lung Res

21:175–185

124. Boyce NW, Campbell D, Holdsworth SR (1989) Granulocyte

independence of pulmonary oxygen toxicity in the rat. Exp Lung

Res 15:491–498

125. Schultz MJ (2008) Lung-protective mechanical ventilation with

lower tidal volumes in patients not suffering from acute lung

injury: a review of clinical studies. Med Sci Monit 14:RA22–

RA26

126. Oeckler RA, Hubmayr RD (2007) Ventilator-associated lung

injury: a search for better therapeutic targets. Eur Respir J

30:1216–1226

127. Thompson BT (2010) Corticosteroids for ARDS. Minerva

Anestesiol 76:441–447

128. Thompson BT (2006) Intensive insulin therapy reduced mor-

bidity but not mortality in patients in the medical intensive care

unit. ACP J Club 145:34

129. Willson DF, Thomas NJ, Markovitz BP et al (2005) Effect of

exogenous surfactant (calfactant) in pediatric acute lung injury:

a randomized controlled trial. JAMA 293:470–476

130. Frank AJ, Thompson BT (2010) Pharmacological treatments for

acute respiratory distress syndrome. Curr Opin Crit Care

16:62–68

NOX enzymes and acute lung injury 2385

123